1
|
Jiang Y, Yuan Y, Qiao G, Deng Z, Liu Z, Zhang Y, Yu L, Lin H, Ma L, Zhang J. Paradoxical action of PP2A inhibition and its potential for therapeutic sensitization. J Cell Physiol 2024:e31413. [PMID: 39150149 DOI: 10.1002/jcp.31413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 07/19/2024] [Accepted: 08/07/2024] [Indexed: 08/17/2024]
Abstract
The protein phosphatase 2A (PP2A), a serine/threonine phosphatase, is recognized as a tumor suppressor involved in diverse cellular processes and essential for maintaining cell viability in vivo. However, endogenous inhibitors of PP2A such as cancerous inhibitor of PP2A (CIP2A) and endogenous nuclear protein inhibitor 2 of PP2A (SET) counteract the anticancer function of PP2A, promoting tumorigenesis, development, and drug resistance in tumors. Surprisingly though, contrary to conventional understanding, inhibition of the tumor suppressor gene PP2A with exogenous small molecule compounds can enhance the efficacy of cancer treatment and achieve superior tumor inhibition. Moreover, exogenous PP2A inhibitors resensitize cancers to treatment and provide novel therapeutic strategies for drug-resistant tumors, which warrant further investigation.
Collapse
Affiliation(s)
- Yue Jiang
- Department of Oncology, Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ying Yuan
- Department of Oncology, Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Guanglei Qiao
- Department of Oncology, Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhoufeng Deng
- Department of Oncology, Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zimei Liu
- Department of Oncology, Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yan Zhang
- Department of Oncology, Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Liping Yu
- Department of Oncology, Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hongjian Lin
- Department of Oncology, Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Lijun Ma
- Department of Oncology, Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jianjun Zhang
- Department of Oncology, Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
2
|
Zhao YX, Ma LB, Yang Z, Wang F, Wang HY, Dang JY. Cancerous inhibitor of protein phosphatase 2A enhances chemoresistance of gastric cancer cells to oxaliplatin. World J Gastrointest Oncol 2023; 15:286-302. [PMID: 36908323 PMCID: PMC9994047 DOI: 10.4251/wjgo.v15.i2.286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/23/2022] [Accepted: 01/05/2023] [Indexed: 02/14/2023] Open
Abstract
BACKGROUND Cancerous inhibitor of protein phosphatase 2A (CIP2A) is a newly discovered oncogene. It is an active cell proliferation regulatory factor that inhibits tumor apoptosis in gastric cancer (GC) cells. CIP2A is functionally related to chemoresistance in various types of tumors according to recent studies. The underlying mechanism, however, is unknown. Further, the primary treatment regimen for GC is oxaliplatin-based chemotherapy. Nonetheless, it often fails due to chemoresistance of GC cells to oxaliplatin.
AIM The goal of this study was to examine CIP2A expression and its association with oxaliplatin resistance in human GC cells.
METHODS Immunohistochemistry was used to examine CIP2A expression in GC tissues and adjacent normal tissues. CIP2A expression in GC cell lines was reduced using small interfering RNA. After confirming the silencing efficiency, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide tetrazolium and flow cytometry assays were used to evaluate cell proliferation and apoptosis caused by oxaliplatin treatment. Further, the key genes and protein changes were verified using real-time quantitative reverse transcription PCR and Western blotting, respectively, before and after intervention. For bioinformatics analysis, we used the R software and Bioconductor project. For statistical analysis, we used GraphPad Prism 6.0 and the Statistical Package for the Social Sciences software version 20.0 (IBM, Armonk, United States).
RESULTS A high level of CIP2A expression was associated with tumor size, T stage, lymph node metastasis, Tumor Node Metastasis stage, and a poor prognosis. Further, CIP2A expression was higher in GC cells than in normal human gastric epithelial cells. Using small interfering RNA against CIP2A, we discovered that CIP2A knockdown inhibited cell proliferation and significantly increased GC cell sensitivity to oxaliplatin. Moreover, CIP2A knockdown enhanced oxaliplatin-induced apoptosis in GC cells. Hence, high CIP2A levels in GC may be a factor in chemoresistance to oxaliplatin. In human GC cells, CIP2A regulated protein kinase B phosphorylation, and chemical inhibition of the protein kinase B signaling pathway was significantly associated with increased sensitivity to oxaliplatin. Therefore, the protein kinase B signaling pathway was correlated with CIP2A-enhanced chemoresistance of human GC cells to oxaliplatin.
CONCLUSION CIP2A expression could be a novel therapeutic strategy for chemoresistance in GC.
Collapse
Affiliation(s)
- Yong-Xun Zhao
- The Seventh Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Li-Bin Ma
- The Seventh Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Ze Yang
- The Seventh Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Fang Wang
- Department of Pathology, The First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Hui-Ying Wang
- The First Clinical Medical School, Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Jia-Yao Dang
- The First Clinical Medical School, Lanzhou University, Lanzhou 730000, Gansu Province, China
| |
Collapse
|
3
|
Chen B, Hu H, Chen X. From Basic Science to Clinical Practice: The Role of Cancerous Inhibitor of Protein Phosphatase 2A (CIP2A)/p90 in Cancer. Front Genet 2023; 14:1110656. [PMID: 36911405 PMCID: PMC9998691 DOI: 10.3389/fgene.2023.1110656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/03/2023] [Indexed: 03/14/2023] Open
Abstract
Cancerous inhibitor of protein phosphatase 2A (CIP2A), initially reported as a tumor-associated antigen (known as p90), is highly expressed in most solid and hematological tumors. The interaction of CIP2A/p90, protein phosphatase 2A (PP2A), and c-Myc can hinder the function of PP2A toward c-Myc S62 induction, thus stabilizing c-Myc protein, which represents a potential role of CIP2A/p90 in tumorigeneses such as cell proliferation, invasion, and migration, as well as cancer drug resistance. The signaling pathways and regulation networks of CIP2A/p90 are complex and not yet fully understood. Many previous studies have also demonstrated that CIP2A/p90 can be used as a potential therapeutic cancer target. In addition, the autoantibody against CIP2A/p90 in sera may be used as a promising biomarker in the diagnosis of certain types of cancer. In this Review, we focus on recent advances relating to CIP2A/p90 and their implications for future research.
Collapse
Affiliation(s)
- Beibei Chen
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China.,Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
| | - Huihui Hu
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China.,Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
| | - Xiaobing Chen
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China.,Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
| |
Collapse
|
4
|
Cristóbal I, Lamarca A. Role of the PP2A Pathway in Cholangiocarcinoma: State of the Art and Future Perspectives. Cancers (Basel) 2022; 14:5422. [PMID: 36358840 PMCID: PMC9657793 DOI: 10.3390/cancers14215422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 10/31/2022] [Indexed: 08/27/2023] Open
Abstract
Cholangiocarcinoma represents a heterogeneous disease at both a clinical and molecular level [...].
Collapse
Affiliation(s)
- Ion Cristóbal
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, IIS-Fundación Jiménez Díaz-UAM, 28040 Madrid, Spain
| | - Angela Lamarca
- Medical Oncology Department, Oncohealth Institute, IIS-Fundación Jiménez Díaz-UAM, 28040 Madrid, Spain
| |
Collapse
|
5
|
Luque M, Cristóbal I, Sanz-Álvarez M, Santos A, Zazo S, Eroles P, Arpí O, Rovira A, Albanell J, Madoz-Gúrpide J, García-Foncillas J, Rojo F. CIP2A as a Key Regulator for AKT Phosphorylation Has Partial Impact Determining Clinical Outcome in Breast Cancer. J Clin Med 2022; 11:jcm11061610. [PMID: 35329936 PMCID: PMC8955826 DOI: 10.3390/jcm11061610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/03/2022] [Accepted: 03/12/2022] [Indexed: 12/07/2022] Open
Abstract
Together with its reported ability to modulate AKT phosphorylation (p-AKT) status in several tumor types, the oncoprotein CIP2A has been described to induce breast cancer progression and drug resistance. However, the clinical and therapeutic relevance of the CIP2A/AKT interplay in breast cancer remains to be fully clarified. Here, we found high p-AKT levels in 80 out of 220 cases (36.4%), which were associated with negative estrogen receptor expression (p = 0.049) and CIP2A overexpression (p < 0.001). Interestingly, p-AKT determined substantially shorter overall (p = 0.002) and progression-free survival (p = 0.003), and multivariate analyses showed its CIP2A-independent prognostic value. Moreover, its clinical relevance was further confirmed in the triple negative and HER2-positive subgroups after stratifying our series by molecular subtype. Functionally, we confirmed in vitro the role of CIP2A as a regulator of p-AKT levels in breast cancer cell lines, and the importance of the CIP2A/AKT axis was also validated in vivo. Finally, p-AKT also showed a higher predictive value of response to doxorubicin than CIP2A in ex vivo analyses. In conclusion, our findings suggest that CIP2A overexpression is a key contributing event to AKT phosphorylation and highlights the CIP2A/AKT axis as a promising therapeutic target in breast cancer. However, our observations highlight the existence of alternative mechanisms that regulate AKT signaling in a subgroup of breast tumors without altered CIP2A expression that determines its independent value as a marker of poor outcome in this disease.
Collapse
Affiliation(s)
- Melani Luque
- Pathology Department, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain; (M.L.); (M.S.-Á.); (S.Z.); (J.M.-G.)
| | - Ion Cristóbal
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, ISS-FJD-UAM, 28040 Madrid, Spain;
- Translational Oncology Division, Oncohealth Institute, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain;
- Correspondence: (I.C.); (F.R.); Tel.: +34-915-504-800 (I.C. & F.R.)
| | - Marta Sanz-Álvarez
- Pathology Department, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain; (M.L.); (M.S.-Á.); (S.Z.); (J.M.-G.)
| | - Andrea Santos
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, ISS-FJD-UAM, 28040 Madrid, Spain;
- Translational Oncology Division, Oncohealth Institute, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain;
| | - Sandra Zazo
- Pathology Department, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain; (M.L.); (M.S.-Á.); (S.Z.); (J.M.-G.)
| | - Pilar Eroles
- Institute of Health Research INCLIVA, 46010 Valencia, Spain;
| | - Oriol Arpí
- Medical Oncology Department, Hospital del Mar, 08003 Barcelona, Spain; (O.A.); (A.R.); (J.A.)
| | - Ana Rovira
- Medical Oncology Department, Hospital del Mar, 08003 Barcelona, Spain; (O.A.); (A.R.); (J.A.)
| | - Joan Albanell
- Medical Oncology Department, Hospital del Mar, 08003 Barcelona, Spain; (O.A.); (A.R.); (J.A.)
| | - Juan Madoz-Gúrpide
- Pathology Department, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain; (M.L.); (M.S.-Á.); (S.Z.); (J.M.-G.)
| | - Jesús García-Foncillas
- Translational Oncology Division, Oncohealth Institute, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain;
| | - Federico Rojo
- Pathology Department, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain; (M.L.); (M.S.-Á.); (S.Z.); (J.M.-G.)
- Correspondence: (I.C.); (F.R.); Tel.: +34-915-504-800 (I.C. & F.R.)
| |
Collapse
|
6
|
Lee HYJ, Meng M, Liu Y, Su T, Kwan HY. Medicinal herbs and bioactive compounds overcome the drug resistance to epidermal growth factor receptor inhibitors in non-small cell lung cancer. Oncol Lett 2021; 22:646. [PMID: 34386068 DOI: 10.3892/ol.2021.12907] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 06/04/2021] [Indexed: 12/12/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related mortality worldwide. Non-small cell lung cancer (NSCLC) accounts for ~85% of all lung cancer cases. Patients harboring epidermal growth factor receptor (EGFR) mutations usually develop resistance to treatment with frontline EGFR-tyrosine kinase inhibitors (EGFR-TKIs). The present review summarizes the current findings and delineates the molecular mechanism of action for the therapeutic effects of herbal extracts and phytochemicals in overcoming EGFR-TKI resistance in NSCLC. Novel molecular targets underlying EGFR-TKI resistance in NSCLC are also discussed. This review provides valuable information for the development of herbal bioactive compounds as alternative treatments for EGFR-TKI-resistant NSCLC.
Collapse
Affiliation(s)
- Hiu Yan Jennifer Lee
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, P.R. China
| | - Mingjing Meng
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Yulong Liu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, P.R. China
| | - Tao Su
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Hiu Yee Kwan
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, P.R. China
| |
Collapse
|
7
|
Saafan H, Alahdab A, Michelet R, Gohlke L, Ziemann J, Holdenrieder S, McLaughlin KM, Wass MN, Cinatl J, Michaelis M, Kloft C, Ritter CA. Constitutive Cell Proliferation Regulating Inhibitor of Protein Phosphatase 2A (CIP2A) Mediates Drug Resistance to Erlotinib in an EGFR Activating Mutated NSCLC Cell Line. Cells 2021; 10:716. [PMID: 33804833 PMCID: PMC8103245 DOI: 10.3390/cells10040716] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 11/21/2022] Open
Abstract
Exploring mechanisms of drug resistance to targeted small molecule drugs is critical for an extended clinical benefit in the treatment of non-small cell lung cancer (NSCLC) patients carrying activating epidermal growth factor receptor (EGFR) mutations. Here, we identified constitutive cell proliferation regulating inhibitor of protein phosphatase 2A (CIP2A) in the HCC4006rErlo0.5 NSCLC cell line adapted to erlotinib as a model of acquired drug resistance. Constitutive CIP2A resulted in a constitutive activation of Akt signaling. The proteasome inhibitor bortezomib was able to reduce CIP2A levels, which resulted in an activation of protein phosphatase 2A and deactivation of Akt. Combination experiments with erlotinib and bortezomib revealed a lack of interaction between the two drugs. However, the effect size of bortezomib was higher in HCC4006rErlo0.5, compared to the erlotinib-sensitive HCC4006 cells, as indicated by an increase in Emax (0.911 (95%CI 0.867-0.954) vs. 0.585 (95%CI 0.568-0.622), respectively) and decrease in EC50 (52.4 µM (95%CI 46.1-58.8 µM) vs. 73.0 µM (95%CI 60.4-111 µM), respectively) in the concentration-effect model, an earlier onset of cell death induction, and a reduced colony surviving fraction (0.38 ± 0.18 vs. 0.95 ± 0.25, respectively, n = 3, p < 0.05). Therefore, modulation of CIP2A with bortezomib could be an interesting approach to overcome drug resistance to erlotinib treatment in NSCLC.
Collapse
Affiliation(s)
- Hisham Saafan
- Institute of Pharmacy, Clinical Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, 17489 Greifswald, Germany; (H.S.); (A.A.); (L.G.)
| | - Ahmad Alahdab
- Institute of Pharmacy, Clinical Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, 17489 Greifswald, Germany; (H.S.); (A.A.); (L.G.)
| | - Robin Michelet
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, 14195 Berlin, Germany; (R.M.); (C.K.)
| | - Linus Gohlke
- Institute of Pharmacy, Clinical Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, 17489 Greifswald, Germany; (H.S.); (A.A.); (L.G.)
| | - Janine Ziemann
- Central Unit for Infection Prevention and Control, University Medicine Greifswald, 17475 Greifswald, Germany;
| | - Stefan Holdenrieder
- Institute of Laboratory Medicine, German Heart Center, Munich Technical University, 80636 Munich, Germany;
| | - Katie-May McLaughlin
- Industrial Biotechnology Centre, School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK; (K.-M.M.); (M.N.W.); (M.M.)
| | - Mark N. Wass
- Industrial Biotechnology Centre, School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK; (K.-M.M.); (M.N.W.); (M.M.)
| | - Jindrich Cinatl
- Institute of Medical Virology, Goethe-University, 60596 Frankfurt am Main, Germany;
| | - Martin Michaelis
- Industrial Biotechnology Centre, School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK; (K.-M.M.); (M.N.W.); (M.M.)
| | - Charlotte Kloft
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, 14195 Berlin, Germany; (R.M.); (C.K.)
| | - Christoph A Ritter
- Institute of Pharmacy, Clinical Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, 17489 Greifswald, Germany; (H.S.); (A.A.); (L.G.)
| |
Collapse
|
8
|
Kim SH, Lee WH, Seong D, An JH, Je HU, Nam HY, Kim SY, Kim SW, Han MW. The role of CIP2A as a therapeutic target of rapamycin in radioresistant head and neck cancer with TP53 mutation. Head Neck 2019; 41:3362-3371. [DOI: 10.1002/hed.25868] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 06/13/2019] [Accepted: 06/18/2019] [Indexed: 02/06/2023] Open
Affiliation(s)
- Song Hee Kim
- Department of OtolaryngologyUlsan University Hospital, University of Ulsan College of Medicine Ulsan Republic of Korea
| | - Won Hyeok Lee
- Department of OtolaryngologyUlsan University Hospital, University of Ulsan College of Medicine Ulsan Republic of Korea
| | - Daseul Seong
- Department of OtolaryngologyUlsan University Hospital, University of Ulsan College of Medicine Ulsan Republic of Korea
| | - Jae Hee An
- Department of OtolaryngologyUlsan University Hospital, University of Ulsan College of Medicine Ulsan Republic of Korea
| | - Hyoung Uk Je
- Department of Radiation OncologyUlsan University Hospital, University of Ulsan College of Medicine Ulsan Republic of Korea
| | - Hae Yun Nam
- Department of Biochemistry and Molecular BiologyAsan Medical Center, University of Ulsan College of Medicine Seoul Republic of Korea
| | - Sang Yoon Kim
- Department of OtolaryngologyAsan Medical Center, University of Ulsan College of Medicine Seoul Republic of Korea
| | - Seong Who Kim
- Department of Biochemistry and Molecular BiologyAsan Medical Center, University of Ulsan College of Medicine Seoul Republic of Korea
| | - Myung Woul Han
- Department of OtolaryngologyUlsan University Hospital, University of Ulsan College of Medicine Ulsan Republic of Korea
- Department of Otolaryngology‐Head and Neck SurgeryLondon Health Sciences Center, Schulich School of Medicine & Dentistry, Western University London Ontario Canada
| |
Collapse
|
9
|
Knockdown of Yin Yang 1 enhances anticancer effects of cisplatin through protein phosphatase 2A-mediated T308 dephosphorylation of AKT. Cell Death Dis 2018; 9:747. [PMID: 29970878 PMCID: PMC6030060 DOI: 10.1038/s41419-018-0774-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 04/23/2018] [Accepted: 05/28/2018] [Indexed: 11/16/2022]
Abstract
Cisplatin is still one of the first-line drugs for chemotherapy of head and neck squamous cell carcinoma (HNSCC) and shows a survival advantage for HNSCC. However, a substantial proportion of HNSCC eventually becomes resistance to cisplatin and the underlying mechanisms remain to be fully understood. Yin Yang 1 (YY1) is a multifunctional protein regulating both gene transcription and protein modifications and also plays a role in chemotherapy resistance. Here, we reported that knockdown of YY1 by lentivirus-mediated short hairpin RNA or tetracycline-inducible short hairpin RNA enhanced cisplatin-induced apoptosis and inhibition of cell proliferation, migration and invasion in the HNSCC cell lines, and inhibition of the xenograft tumor growth. The underlying mechanisms were revealed that knockdown of YY1 downregulated both S473 and T308 phosphorylation of AKT (protein kinase B), which was mainly responsible for cisplatin resistance, whereas overexpression of YY1 upregulated both S473 and T308 phosphorylation. Cisplatin upregulated YY1 mRNA and protein expression and both S473 and T308 phosphorylation of AKT. In the presence of cisplatin, knockdown of YY1 not only blocked cisplatin-induced increase in S473 and T308 phosphorylation of AKT, but still downregulated T308 phosphorylation. Moreover, protein phosphatase 2A (PP2A) antagonist, okadaic acid, upregulated T308, but not S473, phosphorylation, and simultaneously abolished YY1 knockdown-mediated enhancement of cisplatin-induced inhibition of cell proliferation. In addition, knockdown of YY1 promoted PP2A activity through upregulating mRNA and protein expressions of PP2A catalytic subunit alpha (PPP2CA) through the binding of YY1 in the promoter of PPP2CA. Conversely, activating PP2A by forskolin also promoted YY1 degradation and subsequently inhibited T308 phosphorylation. These results suggested that knockdown of YY1 enhanced anticancer effects of cisplatin through PP2A mediating T308 dephosphorylation of AKT, and that targeting YY1 or PP2A would enhance the efficiency of cisplatin chemotherapy in treatment of HNSCC.
Collapse
|
10
|
Zheng HY, Shen FJ, Tong YQ, Li Y. PP2A Inhibits Cervical Cancer Cell Migration by Dephosphorylation of p-JNK, p-p38 and the p-ERK/MAPK Signaling Pathway. Curr Med Sci 2018; 38:115-123. [DOI: 10.1007/s11596-018-1854-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 01/03/2018] [Indexed: 02/06/2023]
|
11
|
Cha G, Xu J, Xu X, Li B, Lu S, Nanding A, Hu S, Liu S. High expression of CIP2A protein is associated with tumor aggressiveness in stage I-III NSCLC and correlates with poor prognosis. Onco Targets Ther 2017; 10:5907-5914. [PMID: 29276394 PMCID: PMC5731340 DOI: 10.2147/ott.s148250] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The aim of this work was to examine the expression of cancerous inhibitor of protein phosphatase 2A (CIP2A) in non-small cell lung cancer (NSCLC) and analyze its correlation with clinical outcomes. CIP2A protein levels were detected by immunohistochemistry (IHC). One hundred and eighty-four of 209 (88.3%) primary stage I-III NSCLC specimens and 4 of 38 (10.5%) adjacent normal lung tissue specimens expressed CIP2A protein. High expression of CIP2A was detected in 38.8% (81/209) of the NSCLC specimens. Patients diagnosed histologically with late-stage NSCLC (p<0.001) and malignant nodes (p=0.001) exhibited high CIP2A expression. Univariate analysis using the log-rank test identified CIP2A expression as a prognostic predictor for overall survival (p=0.005). In multivariate analyses using the Cox regression test, CIP2A expression, T stage, N stage, histological type, and chemotherapy were identified as independent prognostic factors (p=0.007, 0.001, 0.003, <0.001, and <0.001, respectively). Furthermore, Kaplan-Meier survival curves demonstrated that high CIP2A expression indicated poor prognosis in the subgroup of patients with squamous cell carcinoma (p=0.008). Similar results were noted in the subgroup of patients with adenocarcinoma, but the results did not reach statistical significance (p=0.084). We also used univariate analysis and multivariate analysis to assess the prognostic factors for overall survival in the subgroup of patients who received postoperative chemotherapy. CIP2A expression was also an independent prognostic factor in NSCLC patients who received postoperative chemotherapy (p=0.009), along with histological type (p=0.001) and N stage (p=0.034). In conclusion, adding to the accumulating evidence, our research suggested that the CIP2A expression is associated with aggressiveness and correlates with poor prognosis in NSCLC. Our findings also indicated that CIP2A might be a potential therapeutic target against NSCLC.
Collapse
Affiliation(s)
- Geqi Cha
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang
| | - Jianyu Xu
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang
| | - Xiangying Xu
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang
| | - Bin Li
- Department of Plastic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong
| | - Shan Lu
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang
| | - Abiyasi Nanding
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Songliu Hu
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang
| | - Shilong Liu
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang
| |
Collapse
|
12
|
Hyodo T, Ito S, Asano-Inami E, Chen D, Senga T. A regulatory subunit of protein phosphatase 2A, PPP2R5E, regulates the abundance of microtubule crosslinking factor 1. FEBS J 2017; 283:3662-3671. [PMID: 27521566 DOI: 10.1111/febs.13835] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 07/24/2016] [Accepted: 08/12/2016] [Indexed: 12/22/2022]
Abstract
Dynamic changes in microtubule organization are regulated by numerous microtubule-associating proteins and their post-translational modification. Microtubule crosslinking factor 1 (MTCL1) is a recently identified protein that regulates microtubule organization. To obtain further insight into its functions, we searched for proteins that associate with it using mass spectrometry analysis. We found that PPP2R5E, a regulatory subunit of protein phosphatase 2A, interacted with MTCL1. Depletion of PPP2R5E reduced the abundance of MTCL1 abundance, whereas exogenous expression of PPP2R5E increased endogenous MTCL1. Furthermore, inhibition of phosphatase activity by okadaic acid reduced MTCL1, which was restored by the addition of the protease inhibitor MG132. Finally, we show that cells depleted of PPP2R5E and MTCL1 exhibited defects in microtubule organization. Our results suggest that the PPP2R5E phosphatase may contribute to microtubule organization by stabilizing MTCL1.
Collapse
Affiliation(s)
- Toshinori Hyodo
- Division of Cancer Biology, Nagoya University Graduate School of Medicine, Japan
| | - Satoko Ito
- Division of Cancer Biology, Nagoya University Graduate School of Medicine, Japan
| | - Eri Asano-Inami
- Division of Cancer Biology, Nagoya University Graduate School of Medicine, Japan.,Department of Obstetrics and Gynecology Collaborative Research, Bell Research Center, Nagoya University Graduate School of Medicine, Japan
| | - Dan Chen
- Division of Cancer Biology, Nagoya University Graduate School of Medicine, Japan
| | - Takeshi Senga
- Division of Cancer Biology, Nagoya University Graduate School of Medicine, Japan.
| |
Collapse
|
13
|
Velmurugan BK, Lee CH, Chiang SL, Hua CH, Chen MC, Lin SH, Yeh KT, Ko YC. PP2A deactivation is a common event in oral cancer and reactivation by FTY720 shows promising therapeutic potential. J Cell Physiol 2017; 233:1300-1311. [PMID: 28516459 DOI: 10.1002/jcp.26001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 05/09/2017] [Indexed: 12/17/2022]
Abstract
Protein phosphatase 2A (PP2A) is a tumor suppressor gene, that has been frequently deactivated in many types of cancer. However, its molecular and clinical relevance in oral squamous cell carcinoma (OSCC) remain unclear. Here we show that, PP2A deactivation is a common event in oral cancer cells and hyperphosphorylation in its tyrosine-307 (Y307) residue contributes to PP2A deactivation. PP2A restoration by FTY720 treatment reduced cell growth and decreased GSK-3β phosphorylation without significantly altering other PP2A targets. We further detected PP2A phosphorylation in 262 OSCC tissues. Increased expression of p-PP2A in the tumor tissues was significantly correlated with higher N2/N3-stage (aOR = 2.1, 95%CI: 1.2-3.8). Patients with high p-PP2A expression had lower overall survival rates than those with low expression. Hazard ratio analysis showed that, high p-PP2A expression was significantly associated with mortality density (aOR = 2.2, 95%CI: 1.2-4.0) and lower 10-year overall survival (p = 0.027) in lymph node metastasis. However, no interaction was observed between p-PP2A expression and lymph node metastasis. All our results suggest that PP2A is frequently deactivated in oral cancer and determines poor outcome, restoring its expression by FTY720 can be an alternative therapeutic approach in OSCC.
Collapse
Affiliation(s)
- Bharath K Velmurugan
- Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Chien-Hung Lee
- Department of Public Health, College of Health Science, Kaohsiung Medical University, Kaohsuing, Taiwan.,Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shang-Lun Chiang
- Environment-Omics-Diseases Research Center, China Medical University Hospital, Taichung, Taiwan.,Department of Health Risk Management, College of Public Health, China Medical University, Taichung, Taiwan
| | - Chun-Hung Hua
- Department of Otorhinolaryngology, China Medical University Hospital, Taichung, Taiwan
| | - Mei-Chung Chen
- Department of Pathology, Changhua Christian Hospital, Changhua, Taiwan
| | - Shu-Hui Lin
- Department of Pathology, Changhua Christian Hospital, Changhua, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Kun-Tu Yeh
- Department of Pathology, Changhua Christian Hospital, Changhua, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Ying-Chin Ko
- Environment-Omics-Diseases Research Center, China Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
| |
Collapse
|
14
|
Srikar R, Suresh D, Zambre A, Taylor K, Chapman S, Leevy M, Upendran A, Kannan R. Targeted nanoconjugate co-delivering siRNA and tyrosine kinase inhibitor to KRAS mutant NSCLC dissociates GAB1-SHP2 post oncogene knockdown. Sci Rep 2016; 6:30245. [PMID: 27530552 PMCID: PMC4987653 DOI: 10.1038/srep30245] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 07/01/2016] [Indexed: 12/20/2022] Open
Abstract
A tri-block nanoparticle (TBN) comprising of an enzymatically cleavable porous gelatin nanocore encapsulated with gefitinib (tyrosine kinase inhibitor (TKI)) and surface functionalized with cetuximab-siRNA conjugate has been synthesized. Targeted delivery of siRNA to undruggable KRAS mutated non-small cell lung cancer cells would sensitize the cells to TKI drugs and offers an efficient therapy for treating cancer; however, efficient delivery of siRNA and releasing it in cytoplasm remains a major challenge. We have shown TBN can efficiently deliver siRNA to cytoplasm of KRAS mutant H23 Non-Small Cell Lung Cancer (NSCLC) cells for oncogene knockdown; subsequently, sensitizing it to TKI. In the absence of TKI, the nanoparticle showed minimal toxicity suggesting that the cells adapt a parallel GAB1 mediated survival pathway. In H23 cells, activated ERK results in phosphorylation of GAB1 on serine and threonine residues to form GAB1-p85 PI3K complex. In the absence of TKI, knocking down the oncogene dephosphorylated ERK, and negated the complex formation. This event led to tyrosine phosphorylation at Tyr627 domain of GAB1 that regulated EGFR signaling by recruiting SHP2. In the presence of TKI, GAB1-SHP2 dissociation occurs, leading to cell death. The outcome of this study provides a promising platform for treating NSCLC patients harboring KRAS mutation.
Collapse
Affiliation(s)
- R Srikar
- Department of Radiology, Medical Sciences Building, University of Missouri, Columbia, MO 65212, USA
| | - Dhananjay Suresh
- Department of Bioengineering, University of Missouri, Columbia, MO 65212, USA
| | - Ajit Zambre
- Department of Radiology, Medical Sciences Building, University of Missouri, Columbia, MO 65212, USA
| | - Kristen Taylor
- Department of Pathology, Medical Sciences Building, University of Missouri, Columbia, MO 65212, USA
| | - Sarah Chapman
- Notre Dame Integrated Imaging Facility, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Matthew Leevy
- Notre Dame Integrated Imaging Facility, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Anandhi Upendran
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO 65212, USA.,Institute of Clinical and Translational Science, University of Missouri, Columbia, MO 65212, USA
| | - Raghuraman Kannan
- Department of Radiology, Medical Sciences Building, University of Missouri, Columbia, MO 65212, USA.,Department of Bioengineering, University of Missouri, Columbia, MO 65212, USA.,International Center for Nano/Micro Systems and Nanotechnology, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
15
|
Ohama T. [Targeting PP2A inhibitors as a novel anti-cancer strategy
]. Nihon Yakurigaku Zasshi 2015; 145:293-8. [PMID: 26063151 DOI: 10.1254/fpj.145.293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
16
|
Li T, Wang G. Computer-aided targeting of the PI3K/Akt/mTOR pathway: toxicity reduction and therapeutic opportunities. Int J Mol Sci 2014; 15:18856-91. [PMID: 25334061 PMCID: PMC4227251 DOI: 10.3390/ijms151018856] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 09/21/2014] [Accepted: 10/08/2014] [Indexed: 12/14/2022] Open
Abstract
The PI3K/Akt/mTOR pathway plays an essential role in a wide range of biological functions, including metabolism, macromolecular synthesis, cell growth, proliferation and survival. Its versatility, however, makes it a conspicuous target of many pathogens; and the consequential deregulations of this pathway often lead to complications, such as tumorigenesis, type 2 diabetes and cardiovascular diseases. Molecular targeted therapy, aimed at modulating the deregulated pathway, holds great promise for controlling these diseases, though side effects may be inevitable, given the ubiquity of the pathway in cell functions. Here, we review a variety of factors found to modulate the PI3K/Akt/mTOR pathway, including gene mutations, certain metabolites, inflammatory factors, chemical toxicants, drugs found to rectify the pathway, as well as viruses that hijack the pathway for their own synthetic purposes. Furthermore, this evidence of PI3K/Akt/mTOR pathway alteration and related pathogenesis has inspired the exploration of computer-aided targeting of this pathway to optimize therapeutic strategies. Herein, we discuss several possible options, using computer-aided targeting, to reduce the toxicity of molecularly-targeted therapy, including mathematical modeling, to reveal system-level control mechanisms and to confer a low-dosage combination therapy, the potential of PP2A as a therapeutic target, the formulation of parameters to identify patients who would most benefit from specific targeted therapies and molecular dynamics simulations and docking studies to discover drugs that are isoform specific or mutation selective so as to avoid undesired broad inhibitions. We hope this review will stimulate novel ideas for pharmaceutical discovery and deepen our understanding of curability and toxicity by targeting the PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- Tan Li
- Department of Biology, South University of Science and Technology of China, 1088 Xueyuan Rd., Shenzhen 518055, China.
| | - Guanyu Wang
- Department of Biology, South University of Science and Technology of China, 1088 Xueyuan Rd., Shenzhen 518055, China.
| |
Collapse
|